1
|
Arnhold J. Oxidant-Based Cytotoxic Agents During Aging: From Disturbed Energy Metabolism to Chronic Inflammation and Disease Progression. Biomolecules 2025; 15:547. [PMID: 40305309 PMCID: PMC12025200 DOI: 10.3390/biom15040547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 05/02/2025] Open
Abstract
In humans, aging is an inevitable consequence of diminished growth processes after reaching maturity. The high order of biomolecules in cells and tissues is continuously disturbed by numerous physical and chemical destructive impacts. Host-derived oxidant-based cytotoxic agents (reactive species, transition free metal ions, and free heme) contribute considerably to this damage. These agents are under the control of immediately acting antagonizing principles, which are important to ensure cell and tissue homeostasis. In this review, I apply the concept of host-derived cytotoxic agents and their interplay with antagonizing principles to the aging process. During aging, energy metabolism and the supply of tissues with dioxygen and nutrients are increasingly disturbed. In addition, a chronic inflammatory state develops, a condition known as inflammaging. The balance between oxidant-based cytotoxic agents and protective mechanisms is analyzed depending on age-based physiological alterations in ATP production. Disturbances in this balance are associated with the development of age-related diseases and comorbidities. An enhanced production of reactive species from dysfunctional mitochondria, alterations in cellular redox homeostasis, and adaptations to hypoxia are highlighted. Examples of how disturbances between oxidant-based cytotoxic agents and antagonizing principles contribute to the pathogenesis of diseases in persons of advanced age are given.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
2
|
Chen W, Liu D, Lu K, Xu M, Li D, Yan W, Chen S, Li B. Organoids of Musculoskeletal System for Disease Modeling, Drug Screening, and Regeneration. Adv Healthc Mater 2025; 14:e2402444. [PMID: 39610173 DOI: 10.1002/adhm.202402444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/30/2024] [Indexed: 11/30/2024]
Abstract
Musculoskeletal diseases have emerged as the leading cause of disability worldwide, with their prevalence increasing annually. In light of this escalating health challenge, organoids, an emerging technology in tissue engineering, offer promising solutions for disease modeling, drug screening, regeneration, and repair processes. The successful development of musculoskeletal organoids represents a significant breakthrough, providing a novel platform for studying musculoskeletal diseases and facilitating the discovery of new treatments. Moreover, organoids serve as valuable complements to traditional 2D culture methods and animal models, offering rich insights into musculoskeletal biology. This review provides an overview of organoid technology, outlining the construction processes of various musculoskeletal organoids and highlighting their similarities and differences. Furthermore, the challenges associated with organoid technology in musculoskeletal systems are discussed and insights into future perspectives are offered.
Collapse
Affiliation(s)
- Weicheng Chen
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Dachuan Liu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Kai Lu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Mengping Xu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Di Li
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Wei Yan
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Song Chen
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Bin Li
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| |
Collapse
|
3
|
Wang Y, Xu Z, Wei L, Lu Y, Shi Y, Wen S, Lv X, Huang K, Lu F, Qu J, Hu L. KGF-2 Alleviates Dry Eye Disease by Regulating the HMGB1/TLR4 Pathway. Invest Ophthalmol Vis Sci 2025; 66:28. [PMID: 40227178 PMCID: PMC12007672 DOI: 10.1167/iovs.66.4.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
Purpose This study aimed to investigate the protective effects of keratinocyte growth factor-2 (KGF-2) in dry eye disease (DED) and elucidate its mechanism of action through the regulation of the HMGB1/TLR4 pathway. Methods Two in vitro models were established by stimulating hyperosmolar human corneal epithelial cells (HCECs) and RAW 264.7 cells with lipopolysaccharide. A DED mice model was established using scopolamine and an intelligently controlled environmental system. After KGF-2 treatment, the symptoms of the DED mice were assessed. The changes in inflammatory factors were measured using Western blotting and quantitative reverse-transcription polymerase chain reaction (RT-qPCR). RNA sequencing (RNA-seq) was used to identify the key factors involved in KGF-2 treatment, followed by validation through in vivo and in vitro knockdown of the relevant factors. Results KGF-2 treatment significantly relieved DED in the mice model through increased tear secretion, and improved fluorescein staining scores. In addition, the levels of inflammatory factors were effectively lowered in both in vitro and in vivo models. Bulk RNA-seq analysis suggested that KGF-2 exerts its effects by regulating the HMGB1/TLR4 pathway. Furthermore, KGF-2 treatment inhibited the upregulation and nuclear translocation of HMGB1 in the DED model, thereby suppressing the levels of inflammatory factors associated with the HMGB1/TLR4 pathway. Knockdown of HMGB1 in HCECs and glycyrrhizin treatment in DED mice exhibited therapeutic effects similar to those of KGF-2. Conclusions KGF-2 demonstrated protective effects in both in vivo and in vitro DED models by modulating the HMGB1/TLR4 pathway. These findings suggest its potential as a therapeutic agent for DED, warranting further clinical investigation in this regard.
Collapse
Affiliation(s)
- Yuzhou Wang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhiqiang Xu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Linzhi Wei
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Lu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yizhou Shi
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shiyu Wen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiujuan Lv
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kaiyan Huang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fan Lu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jia Qu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Liang Hu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
4
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquivel E, Martinez A, Visser KJ, Joven Araus A, Wang H, Simon A, Yun MH, Del Rio-Tsonis K. Macrophages modulate fibrosis during newt lens regeneration. Stem Cell Res Ther 2024; 15:141. [PMID: 38745238 PMCID: PMC11094960 DOI: 10.1186/s13287-024-03740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Previous studies have suggested that macrophages are present during lens regeneration in newts, but their role in the process is yet to be elucidated. METHODS Here we generated a transgenic reporter line using the newt, Pleurodeles waltl, that traces macrophages during lens regeneration. Furthermore, we assessed early changes in gene expression during lens regeneration using two newt species, Notophthalmus viridescens and Pleurodeles waltl. Finally, we used clodronate liposomes to deplete macrophages during lens regeneration in both species and tested the effect of a subsequent secondary injury after macrophage recovery. RESULTS Macrophage depletion abrogated lens regeneration, induced the formation of scar-like tissue, led to inflammation, decreased iris pigment epithelial cell (iPEC) proliferation, and increased rates of apoptosis in the eye. Some of these phenotypes persisted throughout the last observation period of 100 days and could be attenuated by exogenous FGF2 administration. A distinct transcript profile encoding acute inflammatory effectors was established for the dorsal iris. Reinjury of the newt eye alleviated the effects of macrophage depletion, including the resolution of scar-like tissue, and re-initiated the regeneration process. CONCLUSIONS Together, our findings highlight the importance of macrophages for facilitating a pro-regenerative environment in the newt eye by regulating fibrotic responses, modulating the overall inflammatory landscape, and maintaining the proper balance of early proliferation and late apoptosis of the iPECs.
Collapse
Affiliation(s)
- Georgios Tsissios
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Anthony Sallese
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - J Raul Perez-Estrada
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Jared A Tangeman
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Weihao Chen
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Byran Smucker
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Department of Statistics, Miami University, Oxford, OH, USA
| | - Sophia C Ratvasky
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Erika Grajales-Esquivel
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Arielle Martinez
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Kimberly J Visser
- CRTD/ Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Alberto Joven Araus
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Hui Wang
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Maximina H Yun
- CRTD/ Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH, USA.
- Center for Visual Sciences at, Miami University, Oxford, OH, USA.
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA.
| |
Collapse
|
5
|
Arnhold J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers (Basel) 2023; 16:81. [PMID: 38201509 PMCID: PMC10778456 DOI: 10.3390/cancers16010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammatory processes are related to all stages of tumorigenesis. As inflammation is closely associated with the activation and release of different cytotoxic agents, the interplay between cytotoxic agents and antagonizing principles is highlighted in this review to address the question of how tumor cells overcome the enhanced values of cytotoxic agents in tumors. In tumor cells, the enhanced formation of mitochondrial-derived reactive species and elevated values of iron ions and free heme are antagonized by an overexpression of enzymes and proteins, contributing to the antioxidative defense and maintenance of redox homeostasis. Through these mechanisms, tumor cells can even survive additional stress caused by radio- and chemotherapy. Through the secretion of active agents from tumor cells, immune cells are suppressed in the tumor microenvironment and an enhanced formation of extracellular matrix components is induced. Different oxidant- and protease-based cytotoxic agents are involved in tumor-mediated immunosuppression, tumor growth, tumor cell invasion, and metastasis. Considering the special metabolic conditions in tumors, the main focus here was directed on the disturbed balance between the cytotoxic agents and protective mechanisms in late-stage tumors. This knowledge is mandatory for the implementation of novel anti-cancerous therapeutic approaches.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
6
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquive EL, Martinez A, Visser KJ, Araus AJ, Wang H, Simon A, Yun MH, Rio-Tsonis KD. Macrophages modulate fibrosis during newt lens regeneration. RESEARCH SQUARE 2023:rs.3.rs-3603645. [PMID: 38045376 PMCID: PMC10690311 DOI: 10.21203/rs.3.rs-3603645/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background Previous studies indicated that macrophages play a role during lens regeneration in newts, but their function has not been tested experimentally. Methods Here we generated a transgenic newt reporter line in which macrophages can be visualized in vivo. Using this new tool, we analyzed the location of macrophages during lens regeneration. We uncovered early gene expression changes using bulk RNAseq in two newt species, Notophthalmus viridescens and Pleurodeles waltl. Next, we used clodronate liposomes to deplete macrophages, which inhibited lens regeneration in both newt species. Results Macrophage depletion induced the formation of scar-like tissue, an increased and sustained inflammatory response, an early decrease in iris pigment epithelial cell (iPEC) proliferation and a late increase in apoptosis. Some of these phenotypes persisted for at least 100 days and could be rescued by exogenous FGF2. Re-injury alleviated the effects of macrophage depletion and re-started the regeneration process. Conclusions Together, our findings highlight the importance of macrophages in facilitating a pro-regenerative environment in the newt eye, helping to resolve fibrosis, modulating the overall inflammatory landscape and maintaining the proper balance of early proliferation and late apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Maximina H Yun
- Dresden University of Technology: Technische Universitat Dresden
| | | |
Collapse
|
7
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquivel E, Martinez A, Visser KJ, Araus AJ, Wang H, Simon A, Yun MH, Rio-Tsonis KD. Macrophages modulate fibrosis during newt lens regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.04.543633. [PMID: 37333184 PMCID: PMC10274724 DOI: 10.1101/2023.06.04.543633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Previous studies indicated that macrophages play a role during lens regeneration in newts, but their function has not been tested experimentally. Here we generated a transgenic newt reporter line in which macrophages can be visualized in vivo. Using this new tool, we analyzed the location of macrophages during lens regeneration. We uncovered early gene expression changes using bulk RNAseq in two newt species, Notophthalmus viridescens and Pleurodeles waltl. Next, we used clodronate liposomes to deplete macrophages, which inhibited lens regeneration in both newt species. Macrophage depletion induced the formation of scar-like tissue, an increased and sustained inflammatory response, an early decrease in iris pigment epithelial cell (iPEC) proliferation and a late increase in apoptosis. Some of these phenotypes persisted for at least 100 days and could be rescued by exogenous FGF2. Re-injury alleviated the effects of macrophage depletion and re-started the regeneration process. Together, our findings highlight the importance of macrophages in facilitating a pro-regenerative environment in the newt eye, helping to resolve fibrosis, modulating the overall inflammatory landscape and maintaining the proper balance of early proliferation and late apoptosis.
Collapse
Affiliation(s)
- Georgios Tsissios
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Anthony Sallese
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - J Raul Perez-Estrada
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Jared A Tangeman
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Weihao Chen
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Byran Smucker
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Department of Statistics, Miami University, Oxford, OH, USA
| | - Sophia C Ratvasky
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Erika Grajales-Esquivel
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Arielle Martinez
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Kimberly J Visser
- CRTD Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Alberto Joven Araus
- Karolinska Institute, Department of Cell and Molecular Biology, Stockholm, Sweden
| | - Hui Wang
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Andras Simon
- Karolinska Institute, Department of Cell and Molecular Biology, Stockholm, Sweden
| | - Maximina H Yun
- CRTD Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| |
Collapse
|
8
|
Finburgh EN, Mauduit O, Noguchi T, Bu JJ, Abbas AA, Hakim DF, Bellusci S, Meech R, Makarenkova HP, Afshari NA. Role of FGF10/FGFR2b Signaling in Homeostasis and Regeneration of Adult Lacrimal Gland and Corneal Epithelium Proliferation. Invest Ophthalmol Vis Sci 2023; 64:21. [PMID: 36715672 PMCID: PMC9896866 DOI: 10.1167/iovs.64.1.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Purpose Fibroblast growth factor 10 (FGF10) is involved in eye, meibomian, and lacrimal gland (LG) development, but its function in adult eye structures remains unknown. This study aimed to characterize the role of FGF10 in homeostasis and regeneration of adult LG and corneal epithelium proliferation. Methods Quantitative reverse transcription PCR was used for analysis of FGF10 expression in both early postnatal and adult mouse LG, and RNA sequencing was used to analyze gene expression during LG inflammation. FGF10 was injected into the LG of two mouse models of Sjögren's syndrome and healthy controls. Flow cytometry, BrdU cell proliferation assay, immunostaining, and hematoxylin and eosin staining were used to evaluate the effects of FGF10 injection on inflammation and cell proliferation in vivo. Mouse and human epithelial cell cultures were treated with FGF10 in vitro, and cell viability was assessed using WST-8 and adenosine triphosphate (ATP) quantification assays. Results The level of Fgf10 mRNA expression was lower in adult LG compared to early postnatal LG and was downregulated in chronic inflammation. FGF10 injection into diseased LGs significantly increased cell proliferation and decreased the number of B cells. Mouse and human corneal epithelial cell cultures treated with FGF10 showed significantly higher cell viability and greater cell proliferation. Conclusions FGF10 appears to promote regeneration in damaged adult LGs. These findings have therapeutic potential for developing new treatments for dry eye disease targeting the ability of the cornea and LG to regenerate.
Collapse
Affiliation(s)
- Emma N Finburgh
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| | - Olivier Mauduit
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Takako Noguchi
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| | - Jennifer J Bu
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| | - Anser A Abbas
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| | - Dominic F Hakim
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| | - Saverio Bellusci
- Cardio-Pulmonary Institute and Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University Giessen, Giessen, Germany
| | - Robyn Meech
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Helen P Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Natalie A Afshari
- Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
9
|
Muacevic A, Adler JR, Ghenev P. Fibroblast Growth Factor-2 and the Invasive Potential in Urothelial Malignancies of the Bladder. Cureus 2023; 15:e34147. [PMID: 36843751 PMCID: PMC9949347 DOI: 10.7759/cureus.34147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction Urothelial carcinomas represent a distinct group of malignancies with a high recurrence potential. Multiple studies have established a set of interactions between the tumor cells of urothelial neoplasms and the extracellular matrix regarding invasion and tumor progression. In the present study, we evaluated the expression of fibroblast growth factor-2 (FGF2) in early-stage urothelial carcinomas of the urinary bladder (pTa and pT1) regarding the invasive potential of these tumors. Materials and methods A retrospective non-clinical approach was utilized for the study. Tumor tissue sections used for the initial diagnosis were stained by immunohistochemical means with an anti-FGF2 antibody and the expression within the extracellular matrix was evaluated using a histo-score (h-score). Statistical parameters regarding tumor invasion, FGF2 expression pattern and levels, patient demographic characteristics, and disease recurrence were analyzed for significance. Results A total of 163 cases were analyzed, with an h-score of 110 determined as the optimal cut-off value for invasive potential regarding FGF2 expression, with a sensitivity of 75.4% and a specificity of 78.9%. No statistical correlation was established between the demographic profile of the patients and the occurrence of disease recurrence. Conclusion Based on our results, the study of tumor-extracellular matrix interactions in regards to FGF2 expression is a promising field, at least in urothelial malignancies of the urinary bladder, in regards to tumor invasive potential, while it remains unestablished how these interactions affect metastatic potential.
Collapse
|
10
|
Liu L, Shi Q, Wang K, Qian Y, Zhou L, Bellusci S, Chen C, Dong N. Fibroblast growth factor 10 protects against particulate matter-induced lung injury by inhibiting oxidative stress-mediated pyroptosis via the PI3K/Akt/Nrf2 signaling pathway. Int Immunopharmacol 2022; 113:109398. [PMID: 36461597 DOI: 10.1016/j.intimp.2022.109398] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
Particulate matter (PM) is a major environmental contaminant that causes and worsens respiratory diseases. Fibroblast growth factor 10 (FGF10), a paracrine fibroblast growth factor that specifically stimulates repair and regeneration after injury, has been shown to protect against PM-induced lung injury. However, the underlying mechanisms are still unclear. In this study, the protective effects of FGF10 were investigated using a PM-induced lung injury mouse model in vivo and BEAS-2B cells in vitro. According to the findings, FGF10 treatment alleviated PM-induced oxidative damage and pyroptosis in vivo and in vitro. Mechanistically, FGF10 activated antioxidative Nrf2 signaling. Inhibition of PI3K signaling with LY294002 or Nrf2 signaling with ML385 revealed that FGF10-mediated lung protection was mediated by the PI3K/Akt/Nrf2 pathway. These results collectively indicate that FGF10 inhibits oxidative stress-mediated pyroptosis via the PI3K/Akt/Nrf2 pathway, suggesting a possible therapy for PM-induced lung injury.
Collapse
Affiliation(s)
- Li Liu
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qiangqiang Shi
- Department of Respiratory Medicine, Affiliated Dongyang Hospital of Wenzhou Medical University, Jinhua 322100, China
| | - Kankai Wang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yao Qian
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Liqin Zhou
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Saverio Bellusci
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, 35392 Giessen, Germany.
| | - Chengshui Chen
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Pulmonary and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| | - Nian Dong
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
11
|
Wang Q, Shi Q, Liu L, Qian Y, Dong N. FGF10 mediates protective anti-oxidative effects in particulate matter-induced lung injury through Nrf2 and NF-κB signaling. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1203. [PMID: 36544647 PMCID: PMC9761170 DOI: 10.21037/atm-22-4389] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022]
Abstract
Background Particulate matter (PM), a well-known environmental pollutant, is an independent risk factor associated with the morbidity of various respiratory diseases. Oxidative stress is an important pathophysiological mechanism related to PM exposure, which mediates redox-sensitive inflammatory signaling, leading to lung injury. Fibroblast growth factor 10 (FGF10), a paracrine fibroblast growth factor that mediates mesenchymal to epithelial signaling, participates in epithelial repair during lung injury. However, whether FGF10-mediated repair in PM-induced lung injury is related to the regulation of oxidative stress remains to be elucidated. Methods In vivo, the C57BL/6 mice were randomly divided, with intratracheal instillation of 5 mg/kg FGF10 1 h before 4 mg/kg PM for 2 consecutive days. In vitro, the BEAS-2B cells were pretreated with 10 ng/mL FGF10 before exposed to 200 µg/mL PM. Besides, the specific Nrf2 inhibitor ML385 was adopted in vitro. The harvested lung tissues were pathologic grading scored. The state of oxidative stress was assessed with dihydroethidium (DHE) staining, malondialdehyde (MDA) activity, hydrogen peroxide (H2O2) assays and reactive oxygen species (ROS). The contents of IL-6 and IL-8 in bronchoalveolar lavage (BAL) as well as culture supernatant were quantified by ELISA. The protein levels of nuclear factor erythroid 2 related factor 2 (Nrf2) and nuclear factor-κB (NF-κB) signaling from lung tissue as well as cell lysate were determined by Western blot. Results In this study, recombinant FGF10 administration relieved the degree of lung injury, which is characterized by bronchitis, in a mouse model of PM exposure. In addition, reduced ROS levels, which are indicative of restrained oxidative stress, were also observed. Moreover, two redox-sensitive signaling pathways, Nrf2 and NF-κB, were found to be differentially regulated by FGF10. Using a cellular model of PM exposure, we found that the anti-inflammatory effect of FGF10 on NF-κB signaling was mediated through the regulation of oxidative stress. The anti-oxidative effect relied on the stimulation of Nrf2 signaling. Blockade of Nrf2 signaling with ML385 significantly compromised the anti-inflammatory effect of FGF10. Conclusions These results underscore that the protective anti-oxidative effects of FGF10 in lung injury are mediated by the stimulation of Nrf2 signaling and inhibition of the NF-κB pathway.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Respiratory Medicine, Jinhua Municipal Central Hospital, Jinhua Hospital of Zhejiang University, Jinhua, China;,Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiangqiang Shi
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China;,Department of Respiratory Medicine, Affiliated Dongyang Hospital of Wenzhou Medical University, Jinhua, China
| | - Li Liu
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yao Qian
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Nian Dong
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Mu M, Li B, Zou Y, Wang W, Cao H, Zhang Y, Sun Q, Chen H, Ge D, Tao H, Hu D, Yuan L, Tao X, Wang J. Coal dust exposure triggers heterogeneity of transcriptional profiles in mouse pneumoconiosis and Vitamin D remedies. Part Fibre Toxicol 2022; 19:7. [PMID: 35057792 PMCID: PMC8772169 DOI: 10.1186/s12989-022-00449-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/13/2022] [Indexed: 12/22/2022] Open
Abstract
Background Coal dust particles (CDP), an inevitable by-product of coal mining for the environment, mainly causes coal workers’ pneumoconiosis (CWP). Long-term exposure to coal dust leads to a complex alternation of biological processes during regeneration and repair in the healing lung. However, the cellular and complete molecular changes associated with pulmonary homeostasis caused by respiratory coal dust particles remain unclear. Methods This study mainly investigated the pulmonary toxicity of respirable-sized CDP in mice using unbiased single-cell RNA sequencing. CDP (< 5 μm) collected from the coal mine was analyzed by Scanning Electron Microscope (SEM) and Mass Spectrometer. In addition, western blotting, Elisa, QPCR was used to detect gene expression at mRNA or protein levels. Pathological analysis including HE staining, Masson staining, immunohistochemistry, and immunofluorescence staining were performed to characterize the structure and functional alternation in the pneumoconiosis mouse and verify the reliability of single-cell sequencing results. Results SEM image and Mass Spectrometer analysis showed that coal dust particles generated during coal mine production have been crushed and screened with a diameter of less than 5 µm and contained less than 10% silica. Alveolar structure and pulmonary microenvironment were destroyed, inflammatory and death (apoptosis, autophagy, and necrosis) pathways were activated, leading to pneumoconiosis in post 9 months coal dust stimulation. A distinct abnormally increased alveolar type 2 epithelial cell (AT2) were classified with a highly active state but reduced the antimicrobial-related protein expression of LYZ and Chia1 after CDP exposure. Beclin1, LC3B, LAMP2, TGF-ß, and MLPH were up-regulated induced by CDP, promoting autophagy and pulmonary fibrosis. A new subset of macrophages with M2-type polarization double expressed MLPH + /CD206 + was found in mice having pneumoconiosis but markedly decreased after the Vitamin D treatment. Activated MLPH + /CD206 + M2 macrophages secreted TGF-β1 and are sensitive to Vitamin D treatment. Conclusions This is the first study to reconstruct the pathologic progression and transcriptome pattern of coal pneumoconiosis in mice. Coal dust had obvious toxic effects on lung epithelial cells and macrophages and eventually induced pulmonary fibrosis. CDP-induced M2-type macrophages could be inhibited by VD, which may be related to the alleviation of the pulmonary fibrosis process. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-022-00449-y.
Collapse
|